101
|
Fucà G, Reppel L, Landoni E, Savoldo B, Dotti G. Enhancing Chimeric Antigen Receptor T-Cell Efficacy in Solid Tumors. Clin Cancer Res 2020; 26:2444-2451. [PMID: 32015021 DOI: 10.1158/1078-0432.ccr-19-1835] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/17/2019] [Accepted: 01/29/2020] [Indexed: 12/30/2022]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has been acclaimed as a revolution in cancer treatment following the impressive results in hematologic malignancies. Unfortunately, in patients with solid tumors, objectives responses to CAR T cells are still anecdotal, and important issues are driven by on-target but off-tumor activity of CAR T cells and by the extremely complex biology of solid tumors. Here, we will review the recent attempts to challenge the therapeutic impediments to CAR T-cell therapy in solid tumors. We will focus on the most promising strategies of antigen targeting to improve tumor specificity and address the tumor heterogeneity, efforts to circumvent the physical barriers of the tumor architecture such as subverted tumor vasculature, impediments of CAR T-cell trafficking and immune suppressive microenvironment.
Collapse
Affiliation(s)
- Giovanni Fucà
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina. .,Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Loic Reppel
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Elisa Landoni
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina. .,Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
102
|
Chandran SS, Klebanoff CA. T cell receptor-based cancer immunotherapy: Emerging efficacy and pathways of resistance. Immunol Rev 2020; 290:127-147. [PMID: 31355495 PMCID: PMC7027847 DOI: 10.1111/imr.12772] [Citation(s) in RCA: 200] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 05/09/2019] [Indexed: 12/13/2022]
Abstract
Adoptive cell transfer (ACT) using chimeric antigen receptor (CAR)-modified T cells can induce durable remissions in patients with refractory B-lymphoid cancers. By contrast, results applying CAR-modified T cells to solid malignancies have been comparatively modest. Alternative strategies to redirect T cell specificity and cytolytic function are therefore necessary if ACT is to serve a greater role in human cancer treatments. T cell receptors (TCRs) are antigen recognition structures physiologically expressed by all T cells that have complementary, and in some cases superior, properties to CARs. Unlike CARs, TCRs confer recognition to epitopes derived from proteins residing within any subcellular compartment, including the membrane, cytoplasm and nucleus. This enables TCRs to detect a broad universe of targets, such as neoantigens, cancer germline antigens, and viral oncoproteins. Moreover, because TCRs have evolved to efficiently detect and amplify antigenic signals, these receptors respond to epitope densities many fold smaller than required for CAR-signaling. Herein, we summarize recent clinical data demonstrating that TCR-based immunotherapies can mediate regression of solid malignancies, including immune-checkpoint inhibitor refractory cancers. These trials simultaneously highlight emerging mechanisms of TCR resistance. We conclude by discussing how TCR-based immunotherapies can achieve broader dissemination through innovations in cell manufacturing and non-viral genome integration techniques.
Collapse
Affiliation(s)
- Smita S Chandran
- Center for Cell Engineering and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.,Parker Institute for Cancer Immunotherapy, New York, NY
| | - Christopher A Klebanoff
- Center for Cell Engineering and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.,Parker Institute for Cancer Immunotherapy, New York, NY.,Weill Cornell Medical College, New York, NY
| |
Collapse
|
103
|
Schwab RD, Bedoya DM, King TR, Levine BL, Posey AD. Approaches of T Cell Activation and Differentiation for CAR-T Cell Therapies. Methods Mol Biol 2020; 2086:203-211. [PMID: 31707678 DOI: 10.1007/978-1-0716-0146-4_15] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Chimeric antigen receptor (CAR) T cell therapies are ex vivo manufactured cellular products that have been useful in the treatment of blood cancers and solid tumors. The quality of the final cellular product is influenced by several amenable factors during the manufacturing process. This review discusses several of the influences on cell product phenotype, including the raw starting material, methods of activation and transduction, and culture supplementation.
Collapse
Affiliation(s)
- Robert D Schwab
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Darel Martínez Bedoya
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tiffany R King
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bruce L Levine
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Avery D Posey
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA.
| |
Collapse
|
104
|
Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat Rev Clin Oncol 2019; 17:147-167. [PMID: 31848460 PMCID: PMC7223338 DOI: 10.1038/s41571-019-0297-y] [Citation(s) in RCA: 842] [Impact Index Per Article: 140.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2019] [Indexed: 12/15/2022]
Abstract
T cells genetically engineered to express chimeric antigen receptors (CARs) have proven — and impressive — therapeutic activity in patients with certain subtypes of B cell leukaemia or lymphoma, with promising efficacy also demonstrated in patients with multiple myeloma. Nevertheless, various barriers restrict the efficacy and/or prevent the widespread use of CAR T cell therapies in these patients as well as in those with other cancers, particularly solid tumours. Key challenges relating to CAR T cells include severe toxicities, restricted trafficking to, infiltration into and activation within tumours, suboptimal persistence in vivo, antigen escape and heterogeneity, and manufacturing issues. The evolution of CAR designs beyond the conventional structures will be necessary to address these limitations and to expand the use of CAR T cells to a wider range of malignancies. Investigators are addressing the current obstacles with a wide range of engineering strategies in order to improve the safety, efficacy and applicability of this therapeutic modality. In this Review, we discuss the innovative designs of novel CAR T cell products that are being developed to increase and expand the clinical benefits of these treatments in patients with diverse cancers. Chimeric antigen receptor (CAR) T cell therapy, the first approved therapeutic approach with a genetic engineering component, holds substantial promise in the treatment of a range of cancers but is nevertheless limited by various challenges, including toxicities, intrinsic and acquired resistance mechanisms, and manufacturing issues. In this Review, the authors describe the innovative approaches to the engineering of CAR T cell products that are providing solutions to these challenges and therefore have the potential to considerably improve the safety and effectiveness of treatment. Chimeric antigen receptor (CAR) T cells have induced remarkable responses in patients with certain haematological malignancies, yet various barriers restrict the efficacy and/or prevent the widespread use of this treatment. Investigators are addressing these challenges with engineering strategies designed to improve the safety, efficacy and applicability of CAR T cell therapy. CARs have modular components, and therefore the optimal molecular design of the CAR can be achieved through many variations of the constituent protein domains. Toxicities currently associated with CAR T cell therapy can be mitigated using engineering strategies to make CAR T cells safer and that potentially broaden the range of tumour-associated antigens that can be targeted by overcoming on-target, off-tumour toxicities. CAR T cell efficacy can be enhanced by using engineering strategies to address the various challenges relating to the unique biology of diverse haematological and solid malignancies. Strategies to address the manufacturing challenges can lead to an improved CAR T cell product for all patients.
Collapse
|
105
|
Dafni U, Michielin O, Lluesma SM, Tsourti Z, Polydoropoulou V, Karlis D, Besser MJ, Haanen J, Svane IM, Ohashi PS, Kammula US, Orcurto A, Zimmermann S, Trueb L, Klebanoff CA, Lotze MT, Kandalaft LE, Coukos G. Efficacy of adoptive therapy with tumor-infiltrating lymphocytes and recombinant interleukin-2 in advanced cutaneous melanoma: a systematic review and meta-analysis. Ann Oncol 2019; 30:1902-1913. [PMID: 31566658 DOI: 10.1093/annonc/mdz398] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Adoptive cell therapy (ACT) using autologous tumor-infiltrating lymphocytes (TIL) has been tested in advanced melanoma patients at various centers. We conducted a systematic review and meta-analysis to assess its efficacy on previously treated advanced metastatic cutaneous melanoma. The PubMed electronic database was searched from inception to 17 December 2018 to identify studies administering TIL-ACT and recombinant interleukin-2 (IL-2) following non-myeloablative chemotherapy in previously treated metastatic melanoma patients. Objective response rate (ORR) was the primary end point. Secondary end points were complete response rate (CRR), overall survival (OS), duration of response (DOR) and toxicity. Pooled estimates were derived from fixed or random effect models, depending on the amount of heterogeneity detected. Analysis was carried out separately for high dose (HD) and low dose (LD) IL-2. Sensitivity analyses were carried out. Among 1211 records screened, 13 studies (published 1988 - 2016) were eligible for meta-analysis. Among 410 heavily pretreated patients (some with brain metastasis), 332 received HD-IL-2 and 78 LD-IL-2. The pooled overall ORR estimate was 41% [95% confidence interval (CI) 35% to 48%], and the overall CRR was 12% (95% CI 7% to 16%). For the HD-IL-2 group, the ORR was 43% (95% CI 36% to 50%), while for the LD-IL-2 it was 35% (95% CI 25% to 45%). Corresponding pooled estimates for CRR were 14% (95% CI 7% to 20%) and 7% (95% CI 1% to 12%). The majority of HD-IL-2 complete responders (27/28) remained in remission during the extent of follow-up after CR (median 40 months). Sensitivity analyses yielded similar results. Higher number of infused cells was associated with a favorable response. The ORR for HD-IL-2 compared favorably with the nivolumab/ipilimumab combination following anti-PD-1 failure. TIL-ACT therapy, especially when combined with HD-IL-2, achieves durable clinical benefit and warrants further investigation. We discuss the current position of TIL-ACT in the therapy of advanced melanoma, particularly in the era of immune checkpoint blockade therapy, and review future opportunities for improvement of this approach.
Collapse
Affiliation(s)
- U Dafni
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland; Faculty of Nursing, National and Kapodistrian University of Athens, Athens, Greece
| | - O Michielin
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland
| | - S Martin Lluesma
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Z Tsourti
- Scientific Research Consulting Hellas, Statistics Center, Athens
| | - V Polydoropoulou
- Scientific Research Consulting Hellas, Statistics Center, Athens
| | - D Karlis
- Department of Statistics, Athens University of Economics and Business, Athens, Greece
| | - M J Besser
- Ella Institute for the Treatment and Research of Melanoma and Skin Cancer, Sheba Medical Center, Tel Aviv; Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - J Haanen
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - I-M Svane
- Department of Hematology and Oncology, Center for Cancer Immune Therapy, Herlev Hospital, Herlev, Denmark
| | - P S Ohashi
- Department of Immunology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Canada
| | - U S Kammula
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh
| | - A Orcurto
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland
| | - S Zimmermann
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland
| | - L Trueb
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland
| | - C A Klebanoff
- Center for Cell Engineering and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York; Parker Institute for Cancer Immunotherapy, New York; Weill Cornell Medical College, New York
| | - M T Lotze
- Department of Immunology, University of Pittsburgh Schools of the Health Sciences, Pittsburgh, USA
| | - L E Kandalaft
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - G Coukos
- Department of Oncology, CHUV, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
106
|
Yamamoto TN, Kishton RJ, Restifo NP. Developing neoantigen-targeted T cell-based treatments for solid tumors. Nat Med 2019; 25:1488-1499. [PMID: 31591590 DOI: 10.1038/s41591-019-0596-y] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 08/22/2019] [Indexed: 02/06/2023]
Abstract
Stimulating an immune response against cancer through adoptive transfer of tumor-targeting lymphocytes has shown great promise in hematological malignancies, but clinical efficacy against many common solid epithelial cancers remains low. Targeting 'neoantigens'-the somatic mutations expressed only by tumor cells-might enable tumor destruction without causing undue damage to vital healthy tissues. Major challenges to targeting neoantigens with T cells include heterogeneity and variability in antigen processing and presentation of targets by tumors, and an incomplete understanding of which T cell qualities are essential for clinically effective therapies. Finally, the prospect of targeting somatic tumor mutations to promote T cell destruction of cancer must contend with the biology that not all tumor-expressed 'neoepitopes' actually generate neoantigens that can be functionally recognized and provoke an effective immune response. In this Review, we discuss the promise, progress and challenges for improving neoantigen-targeted T cell-based immunotherapies for cancer.
Collapse
Affiliation(s)
- Tori N Yamamoto
- Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA.,Center for Cell-Based Therapy, NCI, NIH, Bethesda, MD, USA.,Immunology Graduate Group, University of Pennsylvania, Philadelphia, PA, USA
| | - Rigel J Kishton
- Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA.,Center for Cell-Based Therapy, NCI, NIH, Bethesda, MD, USA
| | - Nicholas P Restifo
- Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA. .,Center for Cell-Based Therapy, NCI, NIH, Bethesda, MD, USA. .,Lyell Immunopharma, South San Francisco, CA, USA.
| |
Collapse
|
107
|
Horton BL, Fessenden TB, Spranger S. Tissue Site and the Cancer Immunity Cycle. Trends Cancer 2019; 5:593-603. [PMID: 31706507 PMCID: PMC7521621 DOI: 10.1016/j.trecan.2019.07.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/15/2019] [Accepted: 07/17/2019] [Indexed: 12/22/2022]
Abstract
Checkpoint blockade immunotherapy (CBT) has revolutionized cancer treatment; however, the cellular and molecular factors that govern responsiveness to immunotherapy remain poorly understood. One emerging area of clinical importance is differential responsiveness to CBT across different tissue sites of tumor growth. Each tissue site in the body can contain unique tissue-resident immune cells from both the lymphoid and the myeloid compartment and differences in tissue-specific immune cell composition might predispose tumors in certain tissue sites to be more or less responsive to immunotherapy. Understanding the interplay between tissue-resident and systemic immune responses against tumors will help to determine how to better therapeutically target the immune system to fight cancer. This review summarizes clinical and preclinical investigations of tissue-specific antitumor immune responses and how they influence the tumor immune microenvironment and the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Brendan L Horton
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tim B Fessenden
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Stefani Spranger
- Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA, USA; Biology Department, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
108
|
Springuel L, Lonez C, Alexandre B, Van Cutsem E, Machiels JPH, Van Den Eynde M, Prenen H, Hendlisz A, Shaza L, Carrasco J, Canon JL, Opyrchal M, Odunsi K, Rottey S, Gilham DE, Flament A, Lehmann FF. Chimeric Antigen Receptor-T Cells for Targeting Solid Tumors: Current Challenges and Existing Strategies. BioDrugs 2019; 33:515-537. [PMID: 31363930 PMCID: PMC6790340 DOI: 10.1007/s40259-019-00368-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chimeric antigen receptor-T cells (CAR-Ts) are an exciting new cancer treatment modality exemplified by the recent regulatory approval of two CD19-targeted CAR-T therapies for certain B cell malignancies. However, this success in the hematological setting has yet to translate to a significant level of objective clinical responses in the solid tumor setting. The reason for this lack of translation undoubtedly lies in the substantial challenges raised by solid tumors to all therapies, including CAR-T, that differ from B cell malignancies. For instance, intravenously infused CAR-Ts are likely to make rapid contact with cancerous B cells since both tend to reside in the same vascular compartments within the body. By contrast, solid cancers tend to form discrete tumor masses with an immune-suppressive tumor microenvironment composed of tumor cells and non-tumor stromal cells served by abnormal vasculature that restricts lymphocyte infiltration and suppresses immune function, expansion, and persistence. Moreover, the paucity of uniquely and homogeneously expressed tumor antigens and inherent plasticity of cancer cells provide major challenges to the specificity, potency, and overall effectiveness of CAR-T therapies. This review focuses on the major preclinical and clinical strategies currently being pursued to tackle these challenges in order to drive the success of CAR-T therapy against solid tumors.
Collapse
Affiliation(s)
| | | | | | | | | | - Marc Van Den Eynde
- Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Hans Prenen
- University Hospital Antwerp (UZ Antwerp), Antwerp, Belgium
| | - Alain Hendlisz
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Leila Shaza
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | | - Kunle Odunsi
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | | | | | | |
Collapse
|
109
|
Ajina A, Maher J. Synergistic combination of oncolytic virotherapy with CAR T-cell therapy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 164:217-292. [PMID: 31383406 DOI: 10.1016/bs.pmbts.2019.06.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
For patients with advanced hematological malignancies the therapeutic landscape has been transformed by the emergence of adoptive cell transfer utilizing autologous chimeric antigen receptor (CAR)-redirected T-cells. However, solid tumors have proved far more resistant to this approach. Here, we summarize the numerous challenges faced by CAR T-cells designed to target solid tumors, highlighting, in particular, issues related to impaired trafficking, expansion, and persistence. In parallel, we draw attention to exciting developments in the burgeoning field of oncolytic virotherapy and posit strategies for the synergistic combination of oncolytic viruses with CAR T-cells to improve outcomes for patients with advanced solid tumors.
Collapse
Affiliation(s)
- Adam Ajina
- King's College London, Division of Cancer Studies, Guy's Hospital, London, United Kingdom.
| | - John Maher
- King's College London, Division of Cancer Studies, Guy's Hospital, London, United Kingdom; Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, London, United Kingdom; Department of Immunology, Eastbourne Hospital, East Sussex, United Kingdom
| |
Collapse
|
110
|
Hanada KI, Yu Z, Chappell GR, Park AS, Restifo NP. An effective mouse model for adoptive cancer immunotherapy targeting neoantigens. JCI Insight 2019; 4:124405. [PMID: 31092734 DOI: 10.1172/jci.insight.124405] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 04/17/2019] [Indexed: 12/20/2022] Open
Abstract
The adoptive cell transfer (ACT) of T cells targeting mutated neoantigens can cause objective responses in varieties of metastatic cancers, but the development of new T cell-based treatments relies on accurate animal models. To investigate the therapeutic effect of targeting a neoantigen with ACT, we used T cells from pmel-1 T cell receptor-transgenic mice, known to recognize a WT peptide, gp100, and a mutated version of the peptide that has higher avidity. We gene-engineered B16 cells to express the WT or mutated gp100 epitopes and found that pmel-1-specific T cells targeting a neoantigen tumor target augmented recognition as measured by IFN-γ production. Neoantigen expression by B16 also enhanced the capacity of pmel-1 T cells to trigger the complete and durable regression of large, established, vascularized tumor and required less lymphodepleting conditioning. Targeting neoantigen uncovered the possibility of using enforced expression of the IL-2Rα chain (CD25) in mutation-reactive CD8+ T cells to improve their antitumor functionality. These data reveal that targeting of "mutated-self" neoantigens may lead to improved efficacy and reduced toxicities of T cell-based cellular immunotherapies for patients with cancer.
Collapse
Affiliation(s)
- Ken-Ichi Hanada
- Surgery Branch, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA.,Center for Cell-Based Therapy, NCI, NIH, Bethesda, Maryland, USA
| | - Zhiya Yu
- Surgery Branch, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA.,Center for Cell-Based Therapy, NCI, NIH, Bethesda, Maryland, USA
| | - Gabrielle R Chappell
- Surgery Branch, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA.,Center for Cell-Based Therapy, NCI, NIH, Bethesda, Maryland, USA.,Biomedical Sciences Research Complex, University of St Andrews, North Haugh, St Andrews, United Kingdom
| | - Adam S Park
- Surgery Branch, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA.,Center for Cell-Based Therapy, NCI, NIH, Bethesda, Maryland, USA.,Harvard University, Cambridge, Massachusetts, USA
| | - Nicholas P Restifo
- Surgery Branch, National Cancer Institute (NCI), NIH, Bethesda, Maryland, USA.,Center for Cell-Based Therapy, NCI, NIH, Bethesda, Maryland, USA
| |
Collapse
|
111
|
Hornick EE, Dagvadorj J, Zacharias ZR, Miller AM, Langlois RA, Chen P, Legge KL, Bishop GA, Sutterwala FS, Cassel SL. Dendritic cell NLRC4 regulates influenza A virus-specific CD4 T cell responses through FasL expression. J Clin Invest 2019; 129:2888-2897. [PMID: 31038471 DOI: 10.1172/jci124937] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Influenza A virus (IAV)-specific T cell responses are important correlates of protection during primary and subsequent infections. Generation and maintenance of robust IAV-specific T cell responses relies on T cell interactions with dendritic cells (DCs). In this study, we explore the role of nucleotide-binding domain leucine-rich repeat containing receptor family member NLRC4 in modulating the DC phenotype during IAV infection. Nlrc4-/- mice had worsened survival and increased viral titers during infection, normal innate immune cell recruitment and IAV-specific CD8 T cell responses, but severely blunted IAV-specific CD4 T cell responses compared to wild-type mice. The defect in the pulmonary IAV-specific CD4 T cell response was not a result of defective priming or migration of these cells in Nlrc4-/- mice but was instead due to an increase in FasL+ DCs, resulting in IAV-specific CD4 T cell death. Together, our data support a novel role for NLRC4 in regulating the phenotype of lung DCs during a respiratory viral infection, and thereby influencing the magnitude of protective T cell responses.
Collapse
Affiliation(s)
- Emma E Hornick
- Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Jargalsaikhan Dagvadorj
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Zeb R Zacharias
- Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Ann M Miller
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Ryan A Langlois
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Peter Chen
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Kevin L Legge
- Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Gail A Bishop
- Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Veterans Affairs Medical Center, Iowa City, Iowa, USA
| | - Fayyaz S Sutterwala
- Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Suzanne L Cassel
- Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
112
|
Cellular therapy approaches harnessing the power of the immune system for personalized cancer treatment. Semin Immunol 2019; 42:101306. [DOI: 10.1016/j.smim.2019.101306] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/17/2019] [Indexed: 12/30/2022]
|
113
|
Abstract
Genetically engineered T cells have shown promising activity in the treatment of cancer. However, these cells are also potentially susceptible to immune-suppressive pathways in the tumor microenvironment that may limit their efficacy. In this issue of the JCI, Yamamoto et al. describe a new cellular engineering approach to prevent Fas-mediated inhibition of T cell function, which may be exploited to improve cellular therapy for cancer.
Collapse
|